1
|
Chen P, Cabral H. Enhancing Targeted Drug Delivery through Cell-Specific Endosomal Escape. ChemMedChem 2024; 19:e202400274. [PMID: 38830827 DOI: 10.1002/cmdc.202400274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/05/2024]
Abstract
Endosome is a major barrier in the intracellular delivery of drugs, especially for biologics, such as proteins, peptides, and nucleic acids. After being endocytosed, these cargos will be trapped inside the endosomal compartments and finally degraded in the lysosomes. Thus, various strategies have been developed to facilitate the escape of cargos from the endosomes to improve the intracellular delivery efficiency. While the majority of the studies are focusing on strengthening the endosomal escape capability to maximize the delivery outcome, recent evidence suggests that a careful control of the endosomal escape process could provide opportunity for targeted drug delivery. In this concept review, we examined current delivery systems that can sense intra-endosomal factors or external stimuli for controlling endosomal escape toward a targeted intracellular delivery of cargos. Furthermore, the prospects and challenges of such strategies are discussed.
Collapse
Affiliation(s)
- Pengwen Chen
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Horacio Cabral
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
2
|
De Vleeschauwer SI, van de Ven M, Oudin A, Debusschere K, Connor K, Byrne AT, Ram D, Rhebergen AM, Raeves YD, Dahlhoff M, Dangles-Marie V, Hermans ER. OBSERVE: guidelines for the refinement of rodent cancer models. Nat Protoc 2024; 19:2571-2596. [PMID: 38992214 DOI: 10.1038/s41596-024-00998-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 02/23/2024] [Indexed: 07/13/2024]
Abstract
Existing guidelines on the preparation (Planning Research and Experimental Procedures on Animals: Recommendations for Excellence (PREPARE)) and reporting (Animal Research: Reporting of In Vivo Experiments (ARRIVE)) of animal experiments do not provide a clear and standardized approach for refinement during in vivo cancer studies, resulting in the publication of generic methodological sections that poorly reflect the attempts made at accurately monitoring different pathologies. Compliance with the 3Rs guidelines has mainly focused on reduction and replacement; however, refinement has been harder to implement. The Oncology Best-practices: Signs, Endpoints and Refinements for in Vivo Experiments (OBSERVE) guidelines are the result of a European initiative supported by EurOPDX and INFRAFRONTIER, and aim to facilitate the refinement of studies using in vivo cancer models by offering robust and practical recommendations on approaches to research scientists and animal care staff. We listed cancer-specific clinical signs as a reference point and from there developed sets of guidelines for a wide variety of rodent models, including genetically engineered models and patient derived xenografts. In this Consensus Statement, we systematically and comprehensively address refinement and monitoring approaches during the design and execution of murine cancer studies. We elaborate on the appropriate preparation of tumor-initiating biologicals and the refinement of tumor-implantation methods. We describe the clinical signs to monitor associated with tumor growth, the appropriate follow-up of animals tailored to varying clinical signs and humane endpoints, and an overview of severity assessment in relation to clinical signs, implantation method and tumor characteristics. The guidelines provide oncology researchers clear and robust guidance for the refinement of in vivo cancer models.
Collapse
Affiliation(s)
| | - Marieke van de Ven
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Anaïs Oudin
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Karlijn Debusschere
- Animal Core Facility VUB, Brussels, Belgium
- Core ARTH Animal Facilities, Medicine and Health Sciences Ghent University, Ghent, Belgium
| | - Kate Connor
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Annette T Byrne
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Doreen Ram
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | | | - Maik Dahlhoff
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Els R Hermans
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
3
|
Park SH, Lee JH, Yang SB, Lee DN, Kang TB, Park J. Development of a Peptide-Based Nano-Sized Cathepsin B Inhibitor for Anticancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15041131. [PMID: 37111617 PMCID: PMC10141979 DOI: 10.3390/pharmaceutics15041131] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/17/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023] Open
Abstract
Numerous cathepsin B inhibitors have been developed and are under investigation as potential cancer treatments. They have been evaluated for their ability to inhibit cathepsin B activity and reduce tumor growth. However, they have shown critical limitations, including low anticancer efficacy and high toxicity, due to their low selectivity and delivery problems. In this study, we developed a novel peptide and drug conjugate (PDC)-based cathepsin B inhibitor using cathepsin-B-specific peptide (RR) and bile acid (BA). Interestingly, this RR and BA conjugate (RR–BA) was able to self-assemble in an aqueous solution, and as a result, it formed stable nanoparticles. The nano-sized RR–BA conjugate showed significant cathepsin B inhibitory effects and anticancer effects against mouse colorectal cancer (CT26) cells. Its therapeutic effect and low toxicity were also confirmed in CT26 tumor-bearing mice after intravenous injection. Therefore, based on these results, the RR–BA conjugate could be developed as an effective anticancer drug candidate for inhibiting cathepsin B in anticancer therapy.
Collapse
Affiliation(s)
- So-Hyeon Park
- Department of Applied Life Science, BK21 Program, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Jun-Hyuck Lee
- Department of Applied Life Science, BK21 Program, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Seong-Bin Yang
- Department of Applied Life Science, BK21 Program, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Dong-Nyeong Lee
- Department of Applied Life Science, BK21 Program, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Tae-Bong Kang
- Department of Applied Life Science, BK21 Program, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Jooho Park
- Department of Applied Life Science, BK21 Program, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
- Center for Metabolic Diseases, Konkuk University, Chungju 27478, Republic of Korea
| |
Collapse
|
4
|
Kennedy GT, Holt DE, Azari FS, Bernstein E, Nadeem B, Chang A, Sullivan NT, Segil A, Desphande C, Bensen E, Santini JT, Kucharczuk JC, Delikatny EJ, Bogyo M, Egan AJM, Bradley CW, Eruslanov E, Lickliter JD, Wright G, Singhal S. A Cathepsin-Targeted Quenched Activity-Based Probe Facilitates Enhanced Detection of Human Tumors during Resection. Clin Cancer Res 2022; 28:3729-3741. [PMID: 35792882 DOI: 10.1158/1078-0432.ccr-22-1215] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/02/2022] [Accepted: 07/01/2022] [Indexed: 02/01/2023]
Abstract
PURPOSE Fluorescence-guided surgery using tumor-targeted contrast agents has been developed to improve the completeness of oncologic resections. Quenched activity-based probes that fluoresce after covalently binding to tumor-specific enzymes have been proposed to improve specificity, but none have been tested in humans. Here, we report the successful clinical translation of a cathepsin activity-based probe (VGT-309) for fluorescence-guided surgery. EXPERIMENTAL DESIGN We optimized the specificity, dosing, and timing of VGT-309 in preclinical models of lung cancer. To evaluate clinical feasibility, we conducted a canine study of VGT-309 during pulmonary tumor resection. We then conducted a randomized, double-blind, dose-escalation study in healthy human volunteers receiving VGT-309 to evaluate safety. Finally, we tested VGT-309 in humans undergoing lung cancer surgery. RESULTS In preclinical models, we found highly specific tumor cell labeling that was blocked by a broad spectrum cathepsin inhibitor. When evaluating VGT-309 for guidance during resection of canine tumors, we found that the probe selectively labeled tumors and demonstrated high tumor-to-background ratio (TBR; range: 2.15-3.71). In the Phase I human study, we found that VGT-309 was safe at all doses studied. In the ongoing Phase II trial, we report two cases in which VGT-309 localized visually occult, non-palpable tumors (TBRs = 2.83 and 7.18) in real time to illustrate its successful clinical translation and potential to improve surgical management. CONCLUSIONS This first-in-human study demonstrates the safety and feasibility of VGT-309 to label human pulmonary tumors during resection. These results may be generalizable to other cancers due to cathepsin overexpression in many solid tumors.
Collapse
Affiliation(s)
- Gregory T Kennedy
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - David E Holt
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania
| | - Feredun S Azari
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Elizabeth Bernstein
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Bilal Nadeem
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Ashley Chang
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Neil T Sullivan
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Alix Segil
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Charuhas Desphande
- Department of Pathology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | | | | | - John C Kucharczuk
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Edward J Delikatny
- Department of Radiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Matthew Bogyo
- Department of Pathology, Stanford University, Palo Alto, California
| | - A J Matthew Egan
- Department of Pathology, St. Vincent's Hospital, Melbourne, Australia
| | - Charles W Bradley
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania
| | - Evgeniy Eruslanov
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | | | - Gavin Wright
- Department of Surgery, St. Vincent's Hospital, University of Melbourne, Melbourne, Australia.,Victorian Comprehensive Cancer Centre Alliance, Melbourne, Australia
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
5
|
Preclinical Evaluation of an Activity-Based Probe for Intraoperative Imaging of Esophageal Cancer. Mol Imaging 2022; 2022:5447290. [PMID: 35903245 PMCID: PMC9328188 DOI: 10.1155/2022/5447290] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/23/2022] [Indexed: 11/18/2022] Open
Abstract
Background Early detection and complete resection are important prognostic factors for esophageal cancer (EC). Intraoperative molecular imaging (IMI) using tumor-targeted tracers is effective in many cancer types. However, there are no EC-specific IMI tracers. We sought to test a cathepsin activity-based tracer (VGT-309) for EC resection. Methods Murine (AKR, HNM007) and human (OE19) EC cell lines were screened for cathepsin expression by western blotting. In vitro binding affinity of VGT-309 was evaluated by fluorescence microscopy. Flank tumor models were developed by injecting EC cells into the flanks of BALB/c or athymic nude mice. Mice pretreated with a cathepsin inhibitor (JPM-OEt) were used to confirm on target binding. Animals were injected with 2 mg/kg VGT-309, underwent IMI, and were sacrificed 24 hours after injection. Results Cathepsins B, L, S, and X were expressed by EC cell lines, and all cell lines were labeled in vitro with VGT-309. Fluorescent signal was eliminated when cells were pretreated with JPM-OEt. On biodistribution analysis, VGT-309 accumulated in the liver, kidneys, and spleen without other organ involvement. VGT-309 selectively accumulated in flank allografts and xenografts, with mean signal-to-background ratio of 5.21 (IQR: 4.18-6.73) for flank allografts and 4.34 (IQR: 3.75-5.02) for flank xenografts. Fluorescence microscopy and histopathological analysis confirmed the selective accumulation of the tracer in tumors compared to background normal tissues. Conclusions VGT-309 is an effective tracer for IMI of esophageal cancer. There is potential for clinical translation both as an adjunct to endoscopic detection and for complete removal of disease during esophagectomy.
Collapse
|
6
|
Abstract
Cysteine cathepsins are proteases critical in physiopathological processes and show potential as targets or biomarkers for diseases and medical conditions. The 11 members of the cathepsin family are redundant in some cases but remarkably independent of others, demanding the development of both pan-cathepsin targeting tools as well as probes that are selective for specific cathepsins with little off-target activity. This review addresses the diverse design strategies that have been employed to accomplish this tailored selectivity among cysteine cathepsin targets and the imaging modalities incorporated. The power of these diverse tools is contextualized by briefly highlighting the nature of a few prominent cysteine cathepsins, their involvement in select diseases, and the application of cathepsin imaging probes in research spanning basic biochemical studies to clinical applications.
Collapse
Affiliation(s)
- Kelton A Schleyer
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1345 Center Dr, Gainesville, FL 32610, USA.
| | - Lina Cui
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1345 Center Dr, Gainesville, FL 32610, USA.
| |
Collapse
|
7
|
Turner MA, Amirfakhri S, Nishino H, Lwin TM, Savides TJ, Reid TR, Singer BB, Hoffman RM, Bouvet M. A Patient-Derived Orthotopic Xenograft Model of Gastroesophageal-Junction Adenocarcinoma Translated to the Clinic by Tumor-Targeting Fluorescent Antibodies to Carcinoembryonic-Antigen-Related Cell-Adhesion Molecules. In Vivo 2021; 35:1959-1963. [PMID: 34182469 DOI: 10.21873/invivo.12463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/26/2021] [Accepted: 05/10/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM During surgical resection of gastroesophageal-junction (GEJ) adenocarcinoma, the margin status is often difficult to visualize resulting in high recurrence rates. The aim of the present study was to develop a labelling technique that would allow improved visualization of GEJ tumor margins for surgeons to reduce recurrence rates in a patient-like model. MATERIALS AND METHODS A patient GEJ tumor was obtained from an endoscopic biopsy and implanted subcutaneously in a nude mouse. A patient-derived orthotopic xenograft (PDOX) model was established by implanting tumor fragments grown from a subcutaneous model to the cardia of the stomach of nude mice. CC1/3/5-SAB, an antibody to carcinoembryonic-antigen-related cell-adhesion molecules, was conjugated with infrared dye IRDye800 to create SAB-IR800. Forty-eight hours after i.v. injection of SAB-IR800, GEJ-PDOX mice were imaged. RESULTS Fluorescence imaging with SAB-IR800 brightly visualized the GEJ adenocarcinoma demonstrating specific targeting. In the PDOX model, injection of SAB-IR800 (50 μg) resulted in a tumor to background ratio of 1.78 at 48 hours and 1.86 at 72 hours. CONCLUSION PDOX models of GEJ tumors can be established from patients by endoscopic biopsy without undergoing surgical resection. GEJ PDOX models should be useful for developing novel diagnostics and therapeutics for this recalcitrant disease.
Collapse
Affiliation(s)
- Michael A Turner
- Department of Surgery, University of California San Diego, San Diego, CA, U.S.A.,VA San Diego Healthcare System, San Diego, CA, U.S.A
| | - Siamak Amirfakhri
- Department of Surgery, University of California San Diego, San Diego, CA, U.S.A.,VA San Diego Healthcare System, San Diego, CA, U.S.A
| | - Hiroto Nishino
- Department of Surgery, University of California San Diego, San Diego, CA, U.S.A.,VA San Diego Healthcare System, San Diego, CA, U.S.A
| | - Thinzar M Lwin
- Department of Surgery, University of California San Diego, San Diego, CA, U.S.A
| | - Thomas J Savides
- Department of Medicine, University of California San Diego, San Diego, CA, U.S.A
| | - Tony R Reid
- Department of Medicine, University of California San Diego, San Diego, CA, U.S.A
| | - Bernhard B Singer
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Robert M Hoffman
- Department of Surgery, University of California San Diego, San Diego, CA, U.S.A.,VA San Diego Healthcare System, San Diego, CA, U.S.A.,AntiCancer, Inc., San Diego, CA, U.S.A
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, CA, U.S.A.; .,VA San Diego Healthcare System, San Diego, CA, U.S.A
| |
Collapse
|
8
|
Arora M, Pandey G, Chauhan SS. Cysteine Cathepsins and Their Prognostic and Therapeutic Relevance in Leukemia. ANNALS OF THE NATIONAL ACADEMY OF MEDICAL SCIENCES (INDIA) 2021. [DOI: 10.1055/s-0041-1726151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
AbstractCysteine cathepsins are lysosomal proteases that require Cys-His ion pair in their catalytic site for enzymatic activity. While their aberrant expression and oncogenic functions have been widely reported in solid tumors, recent findings suggest that these proteases also play an important role in the pathogenesis of hematological malignancies. In this review, we summarize the potential clinical implications of cysteine cathepsins as diagnostic and prognostic markers in leukemia, and present evidences which supports the utility of these proteases as potential therapeutic targets in hematological malignancies. We also highlight the available information on the expression patterns, regulation, and potential functions of cysteine cathepsins in normal hematopoiesis and hematological malignancies. In hematopoiesis, cysteine cathepsins play a variety of physiological roles including regulation of hematopoietic stem cell adhesion in the bone marrow, trafficking, and maturation. They are also involved in several functions of immune cells which include the selection of lymphocytes in the thymus, antigen processing, and presentation. However, the expression of cysteine cathepsins is dysregulated in hematological malignancies where they have been shown to play diverse functions. Interestingly, several pieces of evidence over the past few years have demonstrated overexpression of cathepsins in leukemia and their association with worst survival outcomes in patients. Strategies aimed at altering the expression, activity, and subcellular localization of these cathepsins are emerging as potential therapeutic modalaties in the management of hematological malignancies. Recent findings also suggest the involvement of these proteases in modulating the immune response in leukemia and lymphomas.
Collapse
Affiliation(s)
- Mohit Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Garima Pandey
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Shyam S. Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
9
|
Kopansky-Groisman E, Kogan-Zviagin I, Sella-Tavor O, Oron-Herman M, David A. Near-Infrared Fluorescent Activated Polymeric Probe for Imaging Intraluminal Colorectal Cancer Tumors. Biomacromolecules 2019; 20:3547-3556. [PMID: 31381303 DOI: 10.1021/acs.biomac.9b00806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Detection and removal of preneoplastic tumors is crucial for successful colorectal cancer (CRC) therapy. Here we describe the design of a Cathepsin B (CB)-activated polymeric probe, P-(GGFLGK-IR783), for imaging CRC tumors established by intrarectal or subcutaneous (s.c.) implantation of human colon cancer cells (SW-480 and HT-29) in mice. Multiple copies of the near-infrared fluorescent (NIRF) dye IR783 were attached to a single HPMA copolymer backbone via a CB-cleavable linker (GFLG), and the influence of the dye loading on the fluorescence quenching and activation by CB was assessed in vitro, ex vivo, and in vivo. The optimal dose and dosing regimen of P-(GGFLGK-IR783) for colonic tumor detection was determined. Increasing the IR783 loading in the copolymer from 2.5 to 20 mol % resulted in quenching of the fluorescence signal that was activated in vitro by the action of CB from different origins. Following intravenous administration, P-(GGFLGK-IR783)7.5% preferentially accumulated in intrarectal and s.c. implanted tumors, allowing tumor visualization after 4 h and even 48 h postadministration. Activation of P-(GGFLGK-IR783)7.5% by CB was clearly detected in s.c. implanted tumors, revealing about a 4-fold increase in the fluorescence signal in tumors vs healthy colon tissue. The probe containing the CB-cleavable linker produced higher fluorescence signal intensity in tumors, relative to the noncleavable probe. These results indicate that P-(GGFLGK-IR783)7.5% may aid in detecting CRC tumors and can help to guide selective removal of polyps during colonoscopic procedures.
Collapse
Affiliation(s)
- Eva Kopansky-Groisman
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences , Ben-Gurion University of the Negev , Beer-Sheva 84105 , Israel
| | - Inga Kogan-Zviagin
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences , Ben-Gurion University of the Negev , Beer-Sheva 84105 , Israel
| | | | - Mor Oron-Herman
- Advanced Technology Center, Sheba Medical Center , Tel-Hashomer 52621 , Israel
| | - Ayelet David
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences , Ben-Gurion University of the Negev , Beer-Sheva 84105 , Israel
| |
Collapse
|
10
|
Bogdanov AA, Solovyev ID, Savitsky AP. Sensors for Proteolytic Activity Visualization and Their Application in Animal Models of Human Diseases. BIOCHEMISTRY (MOSCOW) 2019; 84:S1-S18. [PMID: 31213192 DOI: 10.1134/s0006297919140013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Various sensors designed for optical and photo(opto)acoustic imaging in living systems are becoming essential components of basic and applied biomedical research. Some of them including those developed for determining enzyme activity in vivo are becoming commercially available. These sensors can be used for various fluorescent signal detection methods: from whole body tomography to endoscopy with miniature cameras. Sensor molecules including enzyme-cleavable macromolecules carrying multiple quenched near-infrared fluorophores are able to deliver their payload in vivo and have long circulation time in bloodstream enabling detection of enzyme activity for extended periods of time at low doses of these sensors. In the future, more effective "activated" probes are expected to become available with optimized sensitivity to enzymatic activity, spectral characteristics suitable for intraoperative imaging of surgical field, biocompatibility and lack of immunogenicity and toxicity. New in vivo optical imaging methods such as the fluorescence lifetime and photo(opto)acoustic imaging will contribute to early diagnosis of human diseases. The use of sensors for in vivo optical imaging will include more extensive preclinical applications of experimental therapies. At the same time, the ongoing development and improvement of optical signal detectors as well as the availability of biologically inert and highly specific fluorescent probes will further contribute to the introduction of fluorescence imaging into the clinic.
Collapse
Affiliation(s)
- A A Bogdanov
- University of Massachusetts Medical School, Department of Radiology, Laboratory of Molecular Imaging Probes, Worcester, MA 01655, USA. .,A. N. Bach Institute of Biochemistry, Federal Research Center "Fundamentals of Biotechnology", Russian Academy of Sciences, Laboratory of Molecular Imaging, Moscow, 119071, Russia.,Lomonosov Moscow State University, Faculty of Bioengineering and Bioinformatics, Moscow, 119991, Russia
| | - I D Solovyev
- A. N. Bach Institute of Biochemistry, Federal Research Center "Fundamentals of Biotechnology", Russian Academy of Sciences, Laboratory of Molecular Imaging, Moscow, 119071, Russia.,A. N. Bach Institute of Biochemistry, Fundamentals of Biotechnology Federal Research Center, Russian Academy of Sciences, Laboratory of Physical Biochemistry, Moscow, 119071, Russia
| | - A P Savitsky
- A. N. Bach Institute of Biochemistry, Federal Research Center "Fundamentals of Biotechnology", Russian Academy of Sciences, Laboratory of Molecular Imaging, Moscow, 119071, Russia.,A. N. Bach Institute of Biochemistry, Fundamentals of Biotechnology Federal Research Center, Russian Academy of Sciences, Laboratory of Physical Biochemistry, Moscow, 119071, Russia
| |
Collapse
|
11
|
Sun N, Wang D, Yao G, Li X, Mei T, Zhou X, Wong KY, Jiang B, Fang Z. pH-dependent and cathepsin B activable CaCO 3 nanoprobe for targeted in vivo tumor imaging. Int J Nanomedicine 2019; 14:4309-4317. [PMID: 31354262 PMCID: PMC6581754 DOI: 10.2147/ijn.s201722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/09/2019] [Indexed: 12/12/2022] Open
Abstract
Background: The intraoperative visualization of tumor cells is a powerful modality for surgical treatment of solid tumors. Since the completeness of tumor excision is closely correlated with the survival of patients, probes that can assist in distinguishing tumor cells are highly demanded. Purpose: In the present study, a fluorescent probe JF1 was synthesized for imaging of tumor cells by conjugating a substrate of cathepsin B (quenching moiety) to Oregon Green derivative JF2 using a self-immolative linker. Methods: JF1 was then loaded into the folate-PEG modified CaCO3 nanoparticles. The folate receptor-targeted, pH-dependent, and cathepsin B activable CaCO3 nanoprobe was test in vitro and in vivo for tumor imaging. Results: CaCO3 nanoprobe demonstrated good stability and fast lighting ability in tumors under low pH conditions. It also showed lower fluorescence background than the single cathepsin B dependent fluorescent probe. The pH-dependent and cathepsin B controlled “turn-on” property enables precise and fast indication of tumor in vitro and in vivo. Conclusion: This strategy of controlled drug delivery enables in vivo imaging of tumor nodules with a high signal-to-noise ratio, which has great potential in surgical tumor treatment.
Collapse
Affiliation(s)
- Ning Sun
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510500, People's Republic of China.,State Key Laboratory of Chemical Biology and Drug Discovery, and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, People's Republic of China
| | - Dou Wang
- Department of Hepatobiliary and Pancreatic Surgery, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen 518020, People's Republic of China
| | - Guoqiang Yao
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, People's Republic of China
| | - Xiaomei Li
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510500, People's Republic of China
| | - Ting Mei
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510500, People's Republic of China
| | - Xinke Zhou
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510500, People's Republic of China
| | - Kwok-Yin Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, People's Republic of China
| | - Baishan Jiang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, People's Republic of China
| | - Zhiyuan Fang
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510500, People's Republic of China
| |
Collapse
|
12
|
Carrier-free nanoparticles of cathepsin B-cleavable peptide-conjugated doxorubicin prodrug for cancer targeting therapy. J Control Release 2018; 294:376-389. [PMID: 30550940 DOI: 10.1016/j.jconrel.2018.11.032] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/24/2018] [Accepted: 11/30/2018] [Indexed: 12/22/2022]
Abstract
Cancer nanomedicine using nanoparticle-based delivery systems has shown outstanding promise in recent decades for improving anticancer treatment. However, limited targeting efficiency, low drug loading efficiency and innate toxicity of nanoparticles have caused severe problems, leaving only a few available in the clinic. Here, we newly developed carrier-free nanoparticles of cathepsin B-cleavable peptide (Phe-Arg-Arg-Gly; FRRG)-conjugated doxorubicin (DOX) prodrug (FRRG-DOX) that formed a stable nanoparticle structure with an average diameter of 213 nm in aqueous condition. The carrier-free nanoparticles of FRRG-DOX induced cytotoxicity against cathepsin B-overexpressed tumor cells whereas the toxicity was minimized in normal cells. In particular, the FRRG-DOX nanoparticles showed the successful tumor-targeting ability and enhanced therapeutic efficiency in human colon adenocarcinoma (HT-29) tumor-bearing mice via enhanced permeation and retention (EPR) effect. Furthermore, FRRG-DOX nanoparticles did not present any severe toxicity, such as non-specific cell death and cardiac toxicity, in normal tissues due to minimal expression of cathepsin B. This carrier-free nanoparticles of FRRG-DOX can solve the unavoidable problems of current nanomedicine, such as lower targeting efficiency, toxicity of nanoparticles themselves, and difficulty in mass production that are fatally caused by natural and synthetic nano-sized carriers.
Collapse
|
13
|
|
14
|
Ding S, Blue RE, Moorefield E, Yuan H, Lund PK. Ex Vivo and In Vivo Noninvasive Imaging of Epidermal Growth Factor Receptor Inhibition on Colon Tumorigenesis Using Activatable Near-Infrared Fluorescent Probes. Mol Imaging 2018; 16:1536012117729044. [PMID: 28884622 PMCID: PMC5595252 DOI: 10.1177/1536012117729044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background: Near-infrared fluorescence (NIRF) imaging combined with enzyme-activatable NIRF probes has yielded promising results in cancer detection. Objective: To test whether 3-dimensional (3-D) noninvasive in vivo NIRF imaging can detect effects of epidermal growth factor receptor (EGFR) inhibitor on both polypoid and flat tumor load in azoxymethane (AOM)-induced colon tumors or tumors in ApcMin/+ mice. Methods: The AOM-injected KK-HIJ mice received EGFR inhibitor diet or chow diet. These and ApcMin/+ mice were given cathepsin-activatable probes (ProSense 680) before imaging. In vivo imaging was performed using quantitative tomographic NIRF imaging. Ex vivo imaging and histologic examination were performed. Dual imaging by micro computed tomography (CT) and 3D NIRF imaging was used to verify tumor location. Results: Tumor load reduction by EGFR inhibition was detected ex vivo using cathepsin B probes. In vivo imaging revealed intense activation of probes only in large tumors. Dual imaging with microCT and 3D NIRF imaging improved tumor detection in vivo. Conclusions: The 3-D NIRF imaging with ProSense 680 can detect and quantify drug effects on colon tumors ex vivo. The NIRF imaging with ProSense 680 probe has limitations as a valid nonendoscopic method for intestinal tumor detection. Combing with other imaging modalities will improve the specificity and sensitivity of intestinal tumor detection in vivo.
Collapse
Affiliation(s)
- Shengli Ding
- 1 Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Randall E Blue
- 1 Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily Moorefield
- 1 Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hong Yuan
- 2 Department of Radiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Pauline K Lund
- 1 Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
15
|
Evaluation of optical imaging agents in a fluorescence-guided surgical model of head and neck cancer. Surg Oncol 2018; 27:225-230. [PMID: 29937175 DOI: 10.1016/j.suronc.2018.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/16/2018] [Accepted: 04/23/2018] [Indexed: 01/23/2023]
Abstract
BACKGROUND Tumor proliferation often occurs from pathologic receptor upregulation. These receptors provide unique targets for near-infrared (NIR) probes that have fluorescence-guided surgery (FGS) applications. We demonstrate the use of three smart-targeted probes in a model of head and neck squamous cell carcinoma. METHODS A dose escalation study was performed using IntegriSense750, ProSense750EX, and ProSense750FAST in mice (n = 5) bearing luciferase-positive SCC-1 flank xenograft tumors. Whole body fluorescence imaging was performed serially after intravenous injection using commercially available open-field (LUNA, Novadaq, Canada) and closed-field NIR systems (Pearl, LI-COR, Lincoln, NE). An ex vivo, whole-body biodistribution was conducted. Lastly, FGS was performed with IntegriSense750 to demonstrate orthotopic and metastatic disease localization. RESULTS Disease fluorescence delineation was assessed by tumor-to-background fluorescence ratios (TBR). Peak TBR values were 3.3 for 1 nmol ProSense750EX, 5.5 for 6 nmol ProSense750FAST, and 10.8 for 4 nmol IntegriSense750 at 5.5, 3, and 4 d post administration, respectively. Agent utility is unique: ProSense750FAST provides sufficient contrast quickly (TBR: 1.5, 3 h) while IntegriSense750 produces strong (TBR: 10.8) contrast with extended administration-to-resection time (96 h). IntegriSense750 correctly identified all diseased nodes in situ during exploratory surgeries. Ex vivo, whole-body biodistribution was assessed by tumor-to-tissue fluorescence ratios (TTR). Agents provided sufficient fluorescence contrast to discriminate disease from background, TTR>1. IntegriSense750 was most robust in neural tissue (TTR: 64) while ProSense750EX was superior localizing disease against lung tissue (TBR: 13). CONCLUSION All three agents appear effective for FGS.
Collapse
|
16
|
Abstract
Cathepsins are lysosomal peptidases belonging to the papain family, and based on their catalytic sites, these enzymes can be divided into serine, cysteine and aspartic proteases. The studies conducted to date have identified, 15 types of cathepsins that are widely distributed in intracellular and extracellular spaces. These proteases participate in various pathological activities, including the occurrence and development of human cancers. Several recent studies suggest that cathepsins, particularly cathepsins B, D, E and L, contribute to digestive tumorigenesis. Cathepsins were found to promote the development of most digestive cancers except liver cancer, in which they might have the opposite effects. Due to their important roles in digestive tumors, cathepsins might be therapeutic targets for the treatment of digestive cancers.
Collapse
|
17
|
Gui L, Yuan Z, Kassaye H, Zheng J, Yao Y, Wang F, He Q, Shen Y, Liang L, Chen H. A tumor-targeting probe based on a mitophagy process for live imaging. Chem Commun (Camb) 2018; 54:9675-9678. [DOI: 10.1039/c8cc04246b] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A pH sensitive probe was designed for mitochondria and autolysosomes monitoring in cell level and tumor targeting imaging.
Collapse
|
18
|
Melsens E, De Vlieghere E, Descamps B, Vanhove C, De Wever O, Ceelen W, Pattyn P. Improved xenograft efficiency of esophageal adenocarcinoma cell lines through in vivo selection. Oncol Rep 2017; 38:71-81. [PMID: 28504813 PMCID: PMC5492849 DOI: 10.3892/or.2017.5640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/18/2016] [Indexed: 01/29/2023] Open
Abstract
The present study aimed to investigate the orthotopic growth potential of two generally available esophageal adenocarcinoma cell lines, OE33 and OACM5 1.C, and a third in vivo selected subpopulation, OACM5 1.C SC1. One group of mice was subcutaneously injected in the hind legs. Tumor growth was measured with calipers. Another group was injected orthotopically in the distal esophageal wall through median laparotomy. Tumor development was evaluated macroscopically and confirmed microscopically. A subset of mice was evaluated with magnetic resonance imaging (MRI) to follow tumor progression. Additionally, functional cell line characteristics were evaluated in vitro (clonogenic, collagen invasion and sphere formation assays, and protein analysis of cell-cell adhesion and cytoskeletal proteins) to better understand xenograft behavior. OE33 cells were shown to be epithelial-like, whereas OACM5 1.C and OACM5 1.C SC1 were more mesenchymal-like. The three cell lines were non-invasive into native type I collagen gels. In vivo, OE33 cells led to 63.6 and 100% tumor nodules after orthotopic (n=12) and subcutaneous (n=8) injection, respectively. Adversely, OACM5 1.C cells did not lead to tumor formation after orthotopic injection (n=6) and only 50% of subcutaneous injections led to tumor nodules (n=8). However, the newly established cell line OACM5 1.C SC1 resulted in 33% tumor formation when orthotopically injected (n=6) and in 100% tumors when injected subcutaneously (n=8). The higher xenograft rate of OACM5 1.C SC1 (P<0.05) corresponded with a higher clonogenic potential compared to its parental cell line (P<0.0001). All models showed local tumor growth without metastasis formation. In conclusion, OACM5 1.C has a poor tumor take rate at an orthotopic and ectopic site. A subpopulation obtained through in vivo selection, OACM5 1.C SC1, gives a significant higher take rate, ectopically. Furthermore, OE33 establishes orthotopic (and subcutaneous) xenografts in mice. These models can be of interest for future studies, and their slow growth rates are a challenge for therapeutic intervention.
Collapse
Affiliation(s)
- Elodie Melsens
- Laboratory of Experimental Surgery, Department of Surgery, Ghent University Hospital, Ghent, Belgium
| | - Elly De Vlieghere
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Benedicte Descamps
- Infinity (iMinds-IBiTech-MEDISIP), Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Christian Vanhove
- Infinity (iMinds-IBiTech-MEDISIP), Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Wim Ceelen
- Laboratory of Experimental Surgery, Department of Surgery, Ghent University Hospital, Ghent, Belgium
| | - Piet Pattyn
- Laboratory of Experimental Surgery, Department of Surgery, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
19
|
Shim MK, Yoon HY, Ryu JH, Koo H, Lee S, Park JH, Kim JH, Lee S, Pomper MG, Kwon IC, Kim K. Cathepsin B-Specific Metabolic Precursor for In Vivo Tumor-Specific Fluorescence Imaging. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201608504] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Man Kyu Shim
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology; 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Republic of Korea
- Department of Pharmacy, Graduate School; Kyung Hee University; 26, Kyungheedae-ro Dongdaemun-gu Seoul 02447 Republic of Korea
| | - Hong Yeol Yoon
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology; 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Republic of Korea
- School of Chemical Engineering; Sungkyunkwan University; 2066, Seobu-ro Jangan-gu Suwon 16419 Republic of Korea
| | - Ju Hee Ryu
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology; 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Republic of Korea
| | - Heebeom Koo
- Department of Medical Life Science, College of Medicine; The Catholic University of Korea; 222, Banpo-daero Seocho-gu Seoul 06591 Republic of Korea
| | - Sangmin Lee
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology; 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Republic of Korea
- The Russell H. Morgan Department of Radiology and Radiological Science; Johns Hopkins University School of Medicine; 601 N. Caroline Street Baltimore MD 21287 USA
| | - Jae Hyung Park
- School of Chemical Engineering; Sungkyunkwan University; 2066, Seobu-ro Jangan-gu Suwon 16419 Republic of Korea
| | - Jong-Ho Kim
- Department of Pharmacy, Graduate School; Kyung Hee University; 26, Kyungheedae-ro Dongdaemun-gu Seoul 02447 Republic of Korea
| | - Seulki Lee
- The Russell H. Morgan Department of Radiology and Radiological Science; Johns Hopkins University School of Medicine; 601 N. Caroline Street Baltimore MD 21287 USA
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science; Johns Hopkins University School of Medicine; 601 N. Caroline Street Baltimore MD 21287 USA
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology; 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology; Korea University; 145 Anam-ro Seongbuk-gu Seoul 02841 Republic of Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute; Korea Institute of Science and Technology; 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Republic of Korea
| |
Collapse
|
20
|
Shim MK, Yoon HY, Ryu JH, Koo H, Lee S, Park JH, Kim J, Lee S, Pomper MG, Kwon IC, Kim K. Cathepsin B‐Specific Metabolic Precursor for In Vivo Tumor‐Specific Fluorescence Imaging. Angew Chem Int Ed Engl 2016; 55:14698-14703. [DOI: 10.1002/anie.201608504] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Indexed: 01/08/2023]
Affiliation(s)
- Man Kyu Shim
- Center for Theragnosis, Biomedical Research Institute Korea Institute of Science and Technology 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Republic of Korea
- Department of Pharmacy, Graduate School Kyung Hee University 26, Kyungheedae-ro Dongdaemun-gu Seoul 02447 Republic of Korea
| | - Hong Yeol Yoon
- Center for Theragnosis, Biomedical Research Institute Korea Institute of Science and Technology 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Republic of Korea
- School of Chemical Engineering Sungkyunkwan University 2066, Seobu-ro Jangan-gu Suwon 16419 Republic of Korea
| | - Ju Hee Ryu
- Center for Theragnosis, Biomedical Research Institute Korea Institute of Science and Technology 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Republic of Korea
| | - Heebeom Koo
- Department of Medical Life Science, College of Medicine The Catholic University of Korea 222, Banpo-daero Seocho-gu Seoul 06591 Republic of Korea
| | - Sangmin Lee
- Center for Theragnosis, Biomedical Research Institute Korea Institute of Science and Technology 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Republic of Korea
- The Russell H. Morgan Department of Radiology and Radiological Science Johns Hopkins University School of Medicine 601 N. Caroline Street Baltimore MD 21287 USA
| | - Jae Hyung Park
- School of Chemical Engineering Sungkyunkwan University 2066, Seobu-ro Jangan-gu Suwon 16419 Republic of Korea
| | - Jong‐Ho Kim
- Department of Pharmacy, Graduate School Kyung Hee University 26, Kyungheedae-ro Dongdaemun-gu Seoul 02447 Republic of Korea
| | - Seulki Lee
- The Russell H. Morgan Department of Radiology and Radiological Science Johns Hopkins University School of Medicine 601 N. Caroline Street Baltimore MD 21287 USA
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science Johns Hopkins University School of Medicine 601 N. Caroline Street Baltimore MD 21287 USA
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute Korea Institute of Science and Technology 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology Korea University 145 Anam-ro Seongbuk-gu Seoul 02841 Republic of Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute Korea Institute of Science and Technology 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Republic of Korea
| |
Collapse
|
21
|
Waterhouse DJ, Joseph J, Neves AA, di Pietro M, Brindle KM, Fitzgerald RC, Bohndiek SE. Design and validation of a near-infrared fluorescence endoscope for detection of early esophageal malignancy. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:84001. [PMID: 27490221 DOI: 10.1117/1.jbo.21.8.084001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/13/2016] [Indexed: 05/24/2023]
Abstract
Barrett’s esophagus is a known precursor lesion to esophageal adenocarcinoma. In these patients, early detection of premalignant disease, known as dysplasia, allows curative minimally invasive endoscopic therapy, but is confounded by a lack of contrast in white light endoscopy. Imaging fluorescently labeled lectins applied topically to the tissue has the potential to more accurately delineate dysplasia, but tissue autofluorescence limits both sensitivity and contrast when operating in the visible region. To overcome this challenge, we synthesized near-infrared (NIR) fluorescent wheat germ agglutinin (WGA-IR800CW) and constructed a clinically translatable bimodal NIR and white light endoscope. Images of NIR and white light with a field of view of 63 deg and an image resolution of 182 μm are coregistered and the honeycomb artifact arising from the fiber bundle is removed. A minimum detectable concentration of 110 nM was determined using a dilution series of WGA-IR800CW. We demonstrated ex vivo that this system can distinguish between gastric and squamous tissue types in mouse stomachs (p=0.0005) and accurately detect WGA-IR800CW fluorescence in human esophageal resections (compared with a gold standard imaging system, rs>0.90). Based on these findings, future work will optimize the bimodal endoscopic system for clinical trials in Barrett’s surveillance.
Collapse
Affiliation(s)
- Dale J Waterhouse
- University of Cambridge, Department of Physics, JJ Thomson Avenue, Cambridge CB3 0HE, United KingdombUniversity of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge CB2 0RE, United Kingdom
| | - James Joseph
- University of Cambridge, Department of Physics, JJ Thomson Avenue, Cambridge CB3 0HE, United KingdombUniversity of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge CB2 0RE, United Kingdom
| | - André A Neves
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge CB2 0RE, United Kingdom
| | - Massimiliano di Pietro
- University of Cambridge, MRC Cancer Unit, Hutchison/MRC Research Centre, Cambridge CB2 0XZ, United Kingdom
| | - Kevin M Brindle
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge CB2 0RE, United KingdomdUniversity of Cambridge, Department of Biochemistry, Sanger Building, Cambridge CB2 1GA, United Kingdom
| | - Rebecca C Fitzgerald
- University of Cambridge, MRC Cancer Unit, Hutchison/MRC Research Centre, Cambridge CB2 0XZ, United Kingdom
| | - Sarah E Bohndiek
- University of Cambridge, Department of Physics, JJ Thomson Avenue, Cambridge CB3 0HE, United KingdombUniversity of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge CB2 0RE, United Kingdom
| |
Collapse
|
22
|
Li Q, Gu W, Liu K, Xiao N, Zhang J, Shao L, Li L, Zhang S, Li P. RGD conjugated, Cy5.5 labeled polyamidoamine dendrimers for targeted near-infrared fluorescence imaging of esophageal squamous cell carcinoma. RSC Adv 2016. [DOI: 10.1039/c6ra12927g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The early detection of esophageal squamous cell carcinoma (ESCC), one of the most common human neoplasms, is of great importance in improving prognosis.
Collapse
Affiliation(s)
- Qi Li
- National Clinical Research Center for Digestive Diseases
- Beijing Digestive Disease Center
- Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases
- Department of Gastroenterology
- Beijing Friendship Hospital
| | - Wei Gu
- School of Chemical Biology and Pharmaceutical Sciences
- Capital Medical University
- People's Republic of China
| | - Kang Liu
- School of Chemical Biology and Pharmaceutical Sciences
- Capital Medical University
- People's Republic of China
| | - Ning Xiao
- Department of Pharmacy
- Beijing Tongren Hospital
- Capital Medical University
- People's Republic of China
| | - Juan Zhang
- School of Chemical Biology and Pharmaceutical Sciences
- Capital Medical University
- People's Republic of China
| | - Linlin Shao
- National Clinical Research Center for Digestive Diseases
- Beijing Digestive Disease Center
- Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases
- Department of Gastroenterology
- Beijing Friendship Hospital
| | - Lei Li
- Department of Gastroenterology
- Affiliated Hospital of Weifang Medical University
- People's Republic of China
| | - Shutian Zhang
- National Clinical Research Center for Digestive Diseases
- Beijing Digestive Disease Center
- Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases
- Department of Gastroenterology
- Beijing Friendship Hospital
| | - Peng Li
- National Clinical Research Center for Digestive Diseases
- Beijing Digestive Disease Center
- Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases
- Department of Gastroenterology
- Beijing Friendship Hospital
| |
Collapse
|
23
|
Sturm MB, Wang TD. Emerging optical methods for surveillance of Barrett's oesophagus. Gut 2015; 64:1816-23. [PMID: 25975605 PMCID: PMC5019028 DOI: 10.1136/gutjnl-2013-306706] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 04/17/2015] [Indexed: 12/20/2022]
Abstract
The rapid rise in incidence of oesophageal adenocarcinoma has motivated the need for improved methods for surveillance of Barrett's oesophagus. Early neoplasia is flat in morphology and patchy in distribution and is difficult to detect with conventional white light endoscopy (WLE). Light offers numerous advantages for rapidly visualising the oesophagus, and advanced optical methods are being developed for wide-field and cross-sectional imaging to guide tissue biopsy and stage early neoplasia, respectively. We review key features of these promising methods and address their potential to improve detection of Barrett's neoplasia. The clinical performance of key advanced imaging technologies is reviewed, including (1) wide-field methods, such as high-definition WLE, chromoendoscopy, narrow-band imaging, autofluorescence and trimodal imaging and (2) cross-sectional techniques, such as optical coherence tomography, optical frequency domain imaging and confocal laser endomicroscopy. Some of these instruments are being adapted for molecular imaging to detect specific biological targets that are overexpressed in Barrett's neoplasia. Gene expression profiles are being used to identify early targets that appear before morphological changes can be visualised with white light. These targets are detected in vivo using exogenous probes, such as lectins, peptides, antibodies, affibodies and activatable enzymes that are labelled with fluorescence dyes to produce high contrast images. This emerging approach has potential to provide a 'red flag' to identify regions of premalignant mucosa, outline disease margins and guide therapy based on the underlying molecular mechanisms of cancer progression.
Collapse
Affiliation(s)
- Matthew B Sturm
- Division of Gastroenterology Departments of Medicine, Biomedical Engineering, Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA,Department of Internal Medicine, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Thomas D Wang
- Division of Gastroenterology Departments of Medicine, Biomedical Engineering, Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA,Departments of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA,Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
24
|
Audran G, Bosco L, Brémond P, Franconi J, Koonjoo N, Marque SRA, Massot P, Mellet P, Parzy E, Thiaudière E. Enzymatically Shifting Nitroxides for EPR Spectroscopy and Overhauser‐Enhanced Magnetic Resonance Imaging. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201506267] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Gérard Audran
- Aix‐Marseille Université, CNRS, ICR, UMR 7273, Case 551, Avenue Escadrille Normandie‐Niemen, 13397 Marseille Cedex 20 (France)
| | - Lionel Bosco
- Aix‐Marseille Université, CNRS, ICR, UMR 7273, Case 551, Avenue Escadrille Normandie‐Niemen, 13397 Marseille Cedex 20 (France)
| | - Paul Brémond
- Aix‐Marseille Université, CNRS, ICR, UMR 7273, Case 551, Avenue Escadrille Normandie‐Niemen, 13397 Marseille Cedex 20 (France)
| | - Jean‐Michel Franconi
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS, Case 93, University Bordeaux Segalen, 146 rue Leo Saignat, 33076 Bordeaux Cedex (France)
| | - Neha Koonjoo
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS, Case 93, University Bordeaux Segalen, 146 rue Leo Saignat, 33076 Bordeaux Cedex (France)
| | - Sylvain R. A. Marque
- Aix‐Marseille Université, CNRS, ICR, UMR 7273, Case 551, Avenue Escadrille Normandie‐Niemen, 13397 Marseille Cedex 20 (France)
| | - Philippe Massot
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS, Case 93, University Bordeaux Segalen, 146 rue Leo Saignat, 33076 Bordeaux Cedex (France)
| | - Philippe Mellet
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS, Case 93, University Bordeaux Segalen, 146 rue Leo Saignat, 33076 Bordeaux Cedex (France)
- INSERM, 33076 Bordeaux Cedex (France)
| | - Elodie Parzy
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS, Case 93, University Bordeaux Segalen, 146 rue Leo Saignat, 33076 Bordeaux Cedex (France)
| | - Eric Thiaudière
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS, Case 93, University Bordeaux Segalen, 146 rue Leo Saignat, 33076 Bordeaux Cedex (France)
| |
Collapse
|
25
|
Audran G, Bosco L, Brémond P, Franconi JM, Koonjoo N, Marque SRA, Massot P, Mellet P, Parzy E, Thiaudière E. Enzymatically Shifting Nitroxides for EPR Spectroscopy and Overhauser-Enhanced Magnetic Resonance Imaging. Angew Chem Int Ed Engl 2015; 54:13379-84. [PMID: 26376730 DOI: 10.1002/anie.201506267] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Indexed: 11/12/2022]
Abstract
In vivo investigations of enzymatic processes using non-invasive approaches are a long-lasting challenge. Recently, we showed that Overhauser-enhanced MRI is suitable to such a purpose. A β-phosphorylated nitroxide substrate prototype exhibiting keto-enol equilibrium upon enzymatic activity has been prepared. Upon enzymatic hydrolysis, a large variation of the phosphorus hyperfine coupling constant (Δa(P)=4 G) was observed. The enzymatic activities of several enzymes were conveniently monitored by electronic paramagnetic resonance (EPR). Using a 0.2 T MRI machine, in vitro and in vivo OMRI experiments were successfully performed, affording a 1200% enhanced MRI signal in vitro, and a 600% enhanced signal in vivo. These results highlight the enhanced imaging potential of these nitroxides upon specific enzymatic substrate-to-product conversion.
Collapse
Affiliation(s)
- Gérard Audran
- Aix-Marseille Université, CNRS, ICR, UMR 7273, Case 551, Avenue Escadrille Normandie-Niemen, 13397 Marseille Cedex 20 (France).
| | - Lionel Bosco
- Aix-Marseille Université, CNRS, ICR, UMR 7273, Case 551, Avenue Escadrille Normandie-Niemen, 13397 Marseille Cedex 20 (France)
| | - Paul Brémond
- Aix-Marseille Université, CNRS, ICR, UMR 7273, Case 551, Avenue Escadrille Normandie-Niemen, 13397 Marseille Cedex 20 (France).
| | - Jean-Michel Franconi
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS, Case 93, University Bordeaux Segalen, 146 rue Leo Saignat, 33076 Bordeaux Cedex (France).
| | - Neha Koonjoo
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS, Case 93, University Bordeaux Segalen, 146 rue Leo Saignat, 33076 Bordeaux Cedex (France)
| | - Sylvain R A Marque
- Aix-Marseille Université, CNRS, ICR, UMR 7273, Case 551, Avenue Escadrille Normandie-Niemen, 13397 Marseille Cedex 20 (France).
| | - Philippe Massot
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS, Case 93, University Bordeaux Segalen, 146 rue Leo Saignat, 33076 Bordeaux Cedex (France)
| | - Philippe Mellet
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS, Case 93, University Bordeaux Segalen, 146 rue Leo Saignat, 33076 Bordeaux Cedex (France). .,INSERM, 33076 Bordeaux Cedex (France).
| | - Elodie Parzy
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS, Case 93, University Bordeaux Segalen, 146 rue Leo Saignat, 33076 Bordeaux Cedex (France)
| | - Eric Thiaudière
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS, Case 93, University Bordeaux Segalen, 146 rue Leo Saignat, 33076 Bordeaux Cedex (France).
| |
Collapse
|
26
|
Özel T, White S, Nguyen E, Moy A, Brenes N, Choi B, Betancourt T. Enzymatically activated near infrared nanoprobes based on amphiphilic block copolymers for optical detection of cancer. Lasers Surg Med 2015; 47:579-594. [PMID: 26189505 DOI: 10.1002/lsm.22396] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2015] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND OBJECTIVE Nanotechnology offers the possibility of creating multi-functional structures that can provide solutions for biomedical problems. The nanoprobes herein described are an example of such structures, where nano-scaled particles have been designed to provide high specificity and contrast potential for optical detection of cancer. Specifically, enzymatically activated fluorescent nanoprobes (EANPs) were synthesized as cancer-specific contrast agents for optical imaging. STUDY DESIGN/MATERIALS AND METHODS EANPs were prepared by nanoprecipitation of blends of poly(lactic acid)-b-poly(ethylene glycol) and poly(lactic-co-glycolic acid)-b-poly(l-lysine). The lysine moieties were then covalently decorated with the near infrared (NIR) fluorescent molecule AlexaFluor-750 (AF750). Close proximity of the fluorescent molecules to each other resulted in fluorescence quenching, which was reversed by enzymatically mediated cleavage of poly(l-lysine) chains. EANPs were characterized by dynamic light scattering and electron microscopy. Enzymatic development of fluorescence was studied in vitro by fluorescence spectroscopy. Biocompatibility and contrast potential of EANPs were studied in cancerous and noncancerous cells. The potential of the nanoprobes as contrast agents for NIR fluorescence imaging was studied in tissue phantoms. RESULTS Spherical EANPs of ∼100 nm were synthesized via nanoprecipitation of polymer blends. Fluorescence activation of EANPs by treatment with a model protease was demonstrated with up to 15-fold optical signal enhancement within 120 minutes. Studies with MDA-MB-231 breast cancer cells demonstrated the cytocompatibility of EANPs, as well as enhanced fluorescence associated with enzymatic activation. Imaging studies in tissue phantoms confirmed the ability of a simple imaging system based on a laser source and CCD camera to image dilute suspensions of the nanoprobe at depths of up to 4 mm, as well as up to a 13-fold signal-to-background ratio for enzymatically activated EANPs compared to un-activated EANPs at the same concentration. CONCLUSION Nanoprecipitation of copolymer blends containing poly(l-lysine) was utilized as a method for preparation of highly functional nanoprobes with high potential as contrast agents for fluorescence based imaging of cancer. Lasers Surg. Med. 47:579-594, 2015. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tuğba Özel
- Materials Science, Engineering, and Commercialization Program, Texas State University, San Marcos, Texas 78666
| | - Sean White
- Department of Biomedical Engineering, Beckman Laser Institute, University of California, Irvine, California 92697
| | - Elaine Nguyen
- Department of Biomedical Engineering, Beckman Laser Institute, University of California, Irvine, California 92697.,School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Austin Moy
- Department of Biomedical Engineering, Beckman Laser Institute, University of California, Irvine, California 92697.,The University of Texas at Austin, Austin, Texas 78712
| | - Nicholas Brenes
- The University of Texas at Austin, Austin, Texas 78712.,InnoSense LLC, Torrance, California 90505
| | - Bernard Choi
- Department of Biomedical Engineering, Beckman Laser Institute, University of California, Irvine, California 92697.,Department of Surgery, University of California, Irvine, California 92697
| | - Tania Betancourt
- Materials Science, Engineering, and Commercialization Program, Texas State University, San Marcos, Texas 78666.,InnoSense LLC, Torrance, California 90505.,Department of Chemistry and Biochemistry, Texas State University San Marcos, Texas 78666
| |
Collapse
|
27
|
Suyama M, Koike M, Asaoka D, Mori H, Oguro M, Ueno T, Nagahara A, Watanabe S, Uchiyama Y. Increased immunoreactivity of cathepsins in the rat esophagus under chronic acid reflux esophagitis. J Histochem Cytochem 2014; 62:645-60. [PMID: 24943348 DOI: 10.1369/0022155414542300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have designed a stable rat chronic acid reflux esophagitis (RE) model. In gastrointestinal lesions, several lysosomal cathepsins are known to participate in epithelial permeability in cell-cell connections, such as tight junctions in ulcerative colitis. However, very few studies have focused on the distribution of cathepsins in the esophageal multilayer squamous epithelium. Therefore to clarify the role of cathepsins in RE, we investigated their immunohistological localization in the esophageal epithelium under normal conditions and after RE. Of the cathepsins examined (cathepsins B, C, D, F, H, L, S, and X), granular immunoreactivity for cathepsins B, C, D and L was observed in the control esophageal epithelia; although, their distribution differed depending on the enzyme examined. In the RE model, immunoreactivity of these cathepsins was increased in esophageal epithelial cells and activated macrophages. The immunoreactivity for cathepsins F, H, S and X was barely detectable in the control esophageal epithelium. However, in the RE model, we noticed a slight increase in the expression of cathepsins H and X in the epithelial cells. Furthermore, activated macrophages of the RE model possessed intense immunoreactivity for these cathepsins, which may have been related to esophageal inflammatory mechanisms.
Collapse
Affiliation(s)
- Masayuki Suyama
- Department of Gastroenterology (MS, DA, HM, MO, AN, SW), Juntendo University School of Medicine, Tokyo, JapanDepartment of Cell Biology and Neuroscience (MK,YU), Juntendo University School of Medicine, Tokyo, JapanCenter for Biomedical Research Resources (TU), Juntendo University Graduate School of Medicine, Tokyo, JapanDepartment of Cellular and Molecular Neuropathology (YU), Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masato Koike
- Department of Gastroenterology (MS, DA, HM, MO, AN, SW), Juntendo University School of Medicine, Tokyo, JapanDepartment of Cell Biology and Neuroscience (MK,YU), Juntendo University School of Medicine, Tokyo, JapanCenter for Biomedical Research Resources (TU), Juntendo University Graduate School of Medicine, Tokyo, JapanDepartment of Cellular and Molecular Neuropathology (YU), Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Daisuke Asaoka
- Department of Gastroenterology (MS, DA, HM, MO, AN, SW), Juntendo University School of Medicine, Tokyo, JapanDepartment of Cell Biology and Neuroscience (MK,YU), Juntendo University School of Medicine, Tokyo, JapanCenter for Biomedical Research Resources (TU), Juntendo University Graduate School of Medicine, Tokyo, JapanDepartment of Cellular and Molecular Neuropathology (YU), Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroki Mori
- Department of Gastroenterology (MS, DA, HM, MO, AN, SW), Juntendo University School of Medicine, Tokyo, JapanDepartment of Cell Biology and Neuroscience (MK,YU), Juntendo University School of Medicine, Tokyo, JapanCenter for Biomedical Research Resources (TU), Juntendo University Graduate School of Medicine, Tokyo, JapanDepartment of Cellular and Molecular Neuropathology (YU), Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masako Oguro
- Department of Gastroenterology (MS, DA, HM, MO, AN, SW), Juntendo University School of Medicine, Tokyo, JapanDepartment of Cell Biology and Neuroscience (MK,YU), Juntendo University School of Medicine, Tokyo, JapanCenter for Biomedical Research Resources (TU), Juntendo University Graduate School of Medicine, Tokyo, JapanDepartment of Cellular and Molecular Neuropathology (YU), Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takashi Ueno
- Department of Gastroenterology (MS, DA, HM, MO, AN, SW), Juntendo University School of Medicine, Tokyo, JapanDepartment of Cell Biology and Neuroscience (MK,YU), Juntendo University School of Medicine, Tokyo, JapanCenter for Biomedical Research Resources (TU), Juntendo University Graduate School of Medicine, Tokyo, JapanDepartment of Cellular and Molecular Neuropathology (YU), Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akihito Nagahara
- Department of Gastroenterology (MS, DA, HM, MO, AN, SW), Juntendo University School of Medicine, Tokyo, JapanDepartment of Cell Biology and Neuroscience (MK,YU), Juntendo University School of Medicine, Tokyo, JapanCenter for Biomedical Research Resources (TU), Juntendo University Graduate School of Medicine, Tokyo, JapanDepartment of Cellular and Molecular Neuropathology (YU), Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Sumio Watanabe
- Department of Gastroenterology (MS, DA, HM, MO, AN, SW), Juntendo University School of Medicine, Tokyo, JapanDepartment of Cell Biology and Neuroscience (MK,YU), Juntendo University School of Medicine, Tokyo, JapanCenter for Biomedical Research Resources (TU), Juntendo University Graduate School of Medicine, Tokyo, JapanDepartment of Cellular and Molecular Neuropathology (YU), Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasuo Uchiyama
- Department of Gastroenterology (MS, DA, HM, MO, AN, SW), Juntendo University School of Medicine, Tokyo, JapanDepartment of Cell Biology and Neuroscience (MK,YU), Juntendo University School of Medicine, Tokyo, JapanCenter for Biomedical Research Resources (TU), Juntendo University Graduate School of Medicine, Tokyo, JapanDepartment of Cellular and Molecular Neuropathology (YU), Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
28
|
Abstract
White light endoscopy has proven to be a very powerful tool in oncology. There is still, however, a need for better endoscopic techniques to overcome the current limitations of white light optics. New technologies that allow higher sensitivity, improved microanatomy and molecular characterization have been available for in vitro microscopy and are now being translated into in vivo endoscopy. Endoscopic molecular imaging is still in its infancy but holds the promise for enhancing sensitivity for early lesions, thus allowing earlier diagnosis and enabling early image-guided endoscopic intervention. A key feature of endoscopic molecular imaging is its increased sensitivity and specificity, which will be illustrated in this article, as well as describing perspectives on its future use in oncologic surgery.
Collapse
Affiliation(s)
- Towhid Ali
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892-1088, USA
| | | | | |
Collapse
|
29
|
Ma W, Ma L, Zhe H, Bao C, Wang N, Yang S, Wang K, Cao F, Cheng Y, Cheng Y. Detection of esophageal squamous cell carcinoma by cathepsin B activity in nude mice. PLoS One 2014; 9:e92351. [PMID: 24618814 PMCID: PMC3950293 DOI: 10.1371/journal.pone.0092351] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 02/21/2014] [Indexed: 02/05/2023] Open
Abstract
Background and Objective Despite great progress in treatment, the prognosis for patients with esophageal squamous cell carcinoma (ESCC) remains poor, highlighting the importance of early detection. Although upper endoscopy can be used for the screening of esophagus, it has limited sensitivity for early stage disease. Thus, development of new diagnosis approach to improve diagnostic capabilities for early detection of ESCC is an important need. The aim of this study was to assess the feasibility of using cathepsin B (CB) as a novel imaging target for the detection of human ESCC by near-infrared optical imaging in nude mice. Methods Initially, we examined specimens from normal human esophageal tissue, intraepithelial neoplasia lesions, tumor in situ, ESCC and two cell lines including one human ESCC cell line (Eca-109) and one normal human esophageal epithelial cell line (HET-1A) for CB expression by immunohistochemistry and western blot, respectively. Next, the ability of a novel CB activatable near-infrared fluorescence (NIRF) probe detecting CB activity presented in Eca-109 cells was confirmed by immunocytochemistry. We also performed in vivo imaging of tumor bearing mice injected with the CB probe and ex vivo imaging of resected tumor xenografts and visceral organs using a living imaging system. Finally, the sources of fluorescence signals in tumor tissue and CB expression in visceral organs were identified by histology. Results CB was absent in normal human esophageal mucosa, but it was overexpressed in ESCC and its precursor lesions. The novel probe for CB activity specifically detected ESCC xenografts in vivo and in vitro. Conclusions CB was highly upregulated in human ESCC and its precursor lesions. The elevated CB expression in ESCC allowed in vivo and in vitro detection of ESCC xenografts in nude mice. Our results support the usefulness of CB activity as a potential imaging target for the detection of human ESCC.
Collapse
Affiliation(s)
- Wei Ma
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
- Department of Radiation Oncology, Cancer Hospital, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Lie Ma
- Department of Cardiology, Cardiovascular and Cerebrovascular Disease Hospital, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Hong Zhe
- Department of Radiation Oncology, Cancer Hospital, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Cihang Bao
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Nana Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Shaoqi Yang
- Digestive System Department, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Kai Wang
- Department of Oncology, Wendeng Center Hospital, Weihai, China
| | - Fangli Cao
- Department of Oncology, Liaocheng People's Hospital, Liaocheng, China
| | - Yanna Cheng
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
- * E-mail:
| |
Collapse
|
30
|
Comparison of multiple enzyme activatable near-infrared fluorescent molecular probes for detection and quantification of inflammation in murine colitis models. Inflamm Bowel Dis 2014; 20:363-77. [PMID: 24374874 PMCID: PMC4618379 DOI: 10.1097/01.mib.0000440612.98950.79] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Activatable near-infrared fluorescent (NIRF) probes have been used for ex vivo and in vivo detection of intestinal tumors in animal models. We hypothesized that NIRF probes activatable by cathepsins or metalloproteinases will detect and quantify dextran sulphate sodium (DSS)-induced acute colonic inflammation in wild type mice or chronic colitis in interleukin-10 (IL-10)-null mice ex vivo or in vivo. METHODS Wild type mice given DSS, water controls, and IL-10-null mice with chronic colitis were administered probes by retro-orbital injection. FMT2500 LX system imaged fresh and fixed intestine ex vivo and mice in vivo. Inflammation detected by probes was verified by histology and colitis scoring. NIRF signal intensity was quantified using 2-dimensional region of interest ex vivo or 3-dimensional region of interest analysis in vivo. RESULTS Ex vivo, 7 probes tested yielded significant higher NIRF signals in colon of DSS-treated mice versus controls. A subset of probes was tested in IL-10-null mice and yielded strong ex vivo signals. Ex vivo fluorescence signal with 680 series probes was preserved after formalin fixation. In DSS and IL-10-null models, ex vivo NIRF signal strongly and significantly correlated with colitis scores. In vivo, ProSense680, CatK680FAST, and MMPsense680 yielded significantly higher NIRF signals in DSS-treated mice than controls, but background was high in controls. CONCLUSIONS Both cathepsin or metalloproteinase-activated NIRF probes can detect and quantify colonic inflammation ex vivo. ProSense680 yielded the strongest signals in DSS colitis ex vivo and in vivo, but background remains a problem for in vivo quantification of colitis.
Collapse
|
31
|
Hu HY, Gehrig S, Reither G, Subramanian D, Mall MA, Plettenburg O, Schultz C. FRET-based and other fluorescent proteinase probes. Biotechnol J 2014; 9:266-81. [PMID: 24464820 DOI: 10.1002/biot.201300201] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Revised: 10/25/2013] [Accepted: 12/24/2013] [Indexed: 12/28/2022]
Abstract
The continuous detection of enzyme activities and their application in medical diagnostics is one of the challenges in the translational sciences. Proteinases represent one of the largest groups of enzymes in the human genome and many diseases are based on malfunctions of proteolytic activity. Fluorescent sensors may shed light on regular and irregular proteinase activity in vitro and in vivo and provide a deeper insight into the function of these enzymes and their role in pathophysiological processes. The focus of this review is on Förster resonance energy transfer (FRET)-based proteinase sensors and reporters because these probes are most likely to provide quantitative data. The medical relevance of proteinases are discussed using lung diseases as a prominent example. Probe design and probe targeting are described and fluorescent probe development for disease-relevant proteinases, including matrix-metalloproteinases, cathepsins, caspases, and other selected proteinases, is reviewed.
Collapse
Affiliation(s)
- Hai-Yu Hu
- European Molecular Biology Laboratory (EMBL), Cell Biology and Biophysics Unit, Heidelberg, Germany; Sanofi Deutschland GmbH, Diabetes Division, R&D, Industriepark Hoechst, Frankfurt am Main, Germany
| | | | | | | | | | | | | |
Collapse
|
32
|
In vivo imaging of tissue-remodeling activity involving infiltration of macrophages by a systemically administered protease-activatable probe in colon cancer tissues. Transl Oncol 2013; 6:628-37. [PMID: 24466365 DOI: 10.1593/tlo.13430] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 11/18/2013] [Accepted: 11/22/2013] [Indexed: 01/16/2023] Open
Abstract
This study evaluated the detection of tumors using in vivo imaging with a commercially available and systemically administered protease-activatable fluorescent probe, ProSense. To this end, we analyzed the delivery and uptake of ProSense as well as the target protease and its cellular source in a mouse xenograft tumor model. In vivo and ex vivo multi wavelength imaging revealed that ProSense signals accumulated within tumors, with preferential distribution in the vascular leakage area that correlates with vasculature development at the tumor periphery. Immunohistochemically, cathepsin B, which is targeted by ProSense, was specifically localized in macrophages. The codistribution of tenascin C immunoreactivity and gelatinase activity provided evidence of tissue-remodeling at the tumor periphery. Furthermore, in situ zymography revealed extracellular ProSense cleavage in such areas. Colocalization of cathepsin B expression and ProSense signals showing reduction by addition of cathepsin B inhibitor was confirmed in cultured macrophage-derived RAW264.7 cells. These results suggest that increased tissue-remodeling activity involving infiltration of macrophages is a mechanism that may be responsible for the tumor accumulation of ProSense signals in our xenograft model. We further confirmed ProSense signals at the tumor margin showing cathepsin B(+) macrophage infiltration in a rat colon carcinogenesis model. Together, these data demonstrate that systemically administered protease-activatable probes can effectively detect cancer invasive fronts, where tissue-remodeling activity is high to facilitate neoplastic cell invasion.
Collapse
|
33
|
Abstract
Molecular imaging is a novel field in gastroenterology that uses fluorescently labelled probes to specifically highlight neoplastic lesions on the basis of their molecular signature. The development of molecular imaging has been driven by the need to improve endoscopic diagnosis and by progress in targeted therapies in gastrointestinal oncology to provide individualized treatment, which coincides with progress in endoscopy techniques and further miniaturization of detection devices. Different exogenous molecular probes for imaging include labelled antibodies, oligopeptides, affibodies(™) (Affibody AB, Bromma, Sweden), aptamers and activatable probes. Molecular imaging has been evaluated in two major indications: many trials have studied molecular imaging as a red flag technique to improve detection of lesions in wide-field imaging; on the other hand, microscopic analysis has been investigated for in vivo characterization of the molecular fingerprint of tumours with the ultimate goal of assessing the likelihood of response to targeted therapy. This Review focusses on the applications of molecular imaging that have immediate potential for translational science or imminent transition into clinical practice of gastrointestinal endoscopy.
Collapse
|
34
|
Cai Y, Zhu L, Zhang F, Niu G, Lee S, Kimura S, Chen X. Noninvasive monitoring of pulmonary fibrosis by targeting matrix metalloproteinases (MMPs). Mol Pharm 2013; 10:2237-47. [PMID: 23607644 PMCID: PMC3672268 DOI: 10.1021/mp300613x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
While idiopathic pulmonary fibrosis (PF) is a devastating lung disease, the management of PF including effective monitoring of disease progression remains a challenge. Herein, we introduce a novel, fast, and ultrasensitive metalloproteinase (MMP) activatable optical probe, named MMP-P12, to noninvasively monitor PF progression and response to PF treatment. A bleomycin (BLM)-induced mouse PF model was subjected noninvasively to optical imaging at various time points after BLM treatment. The mouse PF model developed fibrosis during 21 days of experimental period, and the progression of PF was well correlated with the stepwise increase of MMP-2 expression as examined by quantitative RT-PCR and Western blot analysis on the 7-, 14-, and 21-day post-BLM administration. On these days, MMP-activated fluorescence images were acquired in vivo and ex vivo. Signal quantification showed time-dependent lung-specific incremental increases in fluorescence signals. As a treatment for PF, secretoglobin 3A2 was daily administered intravenously for five days starting on day seven of BLM administration, which resulted in reduced MMP-2 activity and reduction of PF as previously demonstrated. Importantly, the fluorescence signal that reflected MMP activity also decreased in intensity. In conclusion, MMPs may play an important role in PF development and the MMP-P12 probe could be a promising tool for PF detection, even at an early stage of the disease as well as an indicator of therapy response.
Collapse
Affiliation(s)
- Yan Cai
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lei Zhu
- Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361005, China
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fan Zhang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - Seulki Lee
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shioko Kimura
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
35
|
Abstract
Nanotheranostics, the integration of diagnostic and therapeutic function in one system using the benefits of nanotechnology, is extremely attractive for personalized medicine. Because treating cancer is not a one-size-fits-all scenario, it requires therapy to be adapted to the patient's specific biomolecules. Personalized and precision medicine (PM) does just that. It identifies biomarkers to gain an understanding of the diagnosis and in turn treating the specific disorder based on the precise diagnosis. By predominantly utilizing the unique properties of nanoparticles to achieve biomarker identification and drug delivery, nanotheranostics can be applied to noninvasively discover and target image biomarkers and further deliver treatment based on the biomarker distribution. This is a large and hopeful role theranostics must fill. However, as described in this expert opinion, current nanotechnology-based theranostics systems engineered for PM applications are not yet sufficient. PM is an ever-growing field that will be a driving force for future discoveries in biomedicine, especially cancer theranostics. In this article, the authors dissect the requirements for successful nanotheranostics-based PM.
Collapse
Affiliation(s)
- Tae Hyung Kim
- The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Seulki Lee
- The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, NIH, Bethesda, MD, USA
| |
Collapse
|
36
|
Wong GS, Habibollahi P, Heidari P, Lee JS, Klein-Szanto AJ, Waldron TJ, Gimotty P, Nakagawa H, Taylor PR, Wang TC, Mahmood U, Rustgi AK. Optical imaging of periostin enables early endoscopic detection and characterization of esophageal cancer in mice. Gastroenterology 2013; 144:294-297. [PMID: 23085486 PMCID: PMC3624041 DOI: 10.1053/j.gastro.2012.10.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 10/15/2012] [Accepted: 10/16/2012] [Indexed: 12/29/2022]
Abstract
Imaging strategies that detect early stage esophageal squamous cell carcinoma (ESCC) could improve clinical outcomes, when combined with endoscopic approaches. Periostin is an integrin-binding protein that is important in the tumor microenvironment. We created a fluorescent-labeled antibody that recognizes periostin and binds specifically to ESCC xenograft tumors in mice. In L2-cre;p120ctnLoxP/LoxP mice, which develop squamous cell cancers that resemble human ESCC, we visualized the probe in preneoplastic and neoplastic esophageal lesions using near-infrared fluorescent imaging with upper-gastrointestinal endoscopy. Periostin might be a biomarker of the esophageal tumor microenvironment that can be used to detect preneoplastic lesions.
Collapse
Affiliation(s)
- Gabrielle S. Wong
- ) Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Peiman Habibollahi
- ) Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Pedram Heidari
- ) Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Ju-Seog Lee
- ) Department of Systems Biology, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Andres J. Klein-Szanto
- ) Department of Pathology and Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, 19104, USA
| | - Todd J. Waldron
- ) Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Phyllis Gimotty
- ) Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA,) Division of Biostatistics, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hiroshi Nakagawa
- ) Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Philip R. Taylor
- ) Genetic Epidemiology Branch, DCEG, NCI, NIH, Bethesda, MD 20892, USA
| | - Timothy C. Wang
- ) Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA,) Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, 10032, USA
| | - Umar Mahmood
- ) Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Anil K. Rustgi
- ) Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA,) Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
37
|
Bogdanov AA, Mazzanti ML. Fluorescent macromolecular sensors of enzymatic activity for in vivo imaging. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 113:349-87. [PMID: 23244795 DOI: 10.1016/b978-0-12-386932-6.00009-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Macromolecular imaging probes (or sensors) of enzymatic activity have a unique place in the armamentarium of modern optical imaging techniques. Such probes were initially developed by attaching optically "silent" fluorophores via enzyme-sensitive linkers to large copolymers of biocompatible poly(ethylene glycol) and poly(amino acids). In diseased tissue, where the concentration of enzymes is high, the fluorophores are freed from the macromolecular carrier and regain their initial ability to fluoresce, thus allowing in vivo optical localization of the diseased tissue. This chapter describes the design and application of these probes and their alternatives in various areas of experimental medicine and gives an overview of currently available techniques that allow imaging of animals using visible and near-infrared light.
Collapse
Affiliation(s)
- Alexei A Bogdanov
- Laboratory of Molecular Imaging Probes, Department of Radiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | |
Collapse
|
38
|
Lee S, Kim K. Protease activity: meeting its theranostic potential. Theranostics 2012; 2:125-6. [PMID: 22400061 PMCID: PMC3296469 DOI: 10.7150/thno.4129] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Accepted: 01/21/2012] [Indexed: 12/19/2022] Open
Abstract
This themed issue provides up-to-date review and research articles covering the theranostic applications in the combined fields of protease research, diagnostics and drug development.
Collapse
|